Jurnal 5

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/342502492

Minority report: the intestinal mycobiota in systemic infections

Article  in  Current Opinion in Microbiology · August 2020


DOI: 10.1016/j.mib.2020.05.004

CITATIONS READS

2 45

3 authors, including:

Tobias M Hohl
Memorial Sloan Kettering Cancer Center
135 PUBLICATIONS   6,309 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Initiation of the Immune Response against Aspergillus fumigatus View project

All content following this page was uploaded by Tobias M Hohl on 15 September 2020.

The user has requested enhancement of the downloaded file.


Available online at www.sciencedirect.com

ScienceDirect

Minority report: the intestinal mycobiota in systemic


infections
Thierry Rolling1,2,3, Tobias M Hohl1,2 and Bing Zhai1,2,3

Compared to bacteria, fungi often exhibit a lower abundance developments that link the mycobiota to various human
and a higher temporal volatility in the intestinal microbiota. infectious diseases.
Analysis of fungi in the microbiota (mycobiota) faces technical
limitations with tools that were originally developed for Demographics of the human mycobiota
analyzing bacteria. Dysbiotic states of the intestinal mycobiota, Current analytical tools for microbiota largely rely on
often associated with disruption of the healthy bacterial next-generation sequencing techniques and were initially
microbiota, are characterized by overgrowth (domination) of developed for analyzing bacteria. These tools have been
specific fungal taxa and loss of diversity. Intestinal domination adapted to explore the demographics of the mycobiota.
by Candida species has been shown to be a major source of Internal transcribed spacer 1 and 2 (ITS1 and ITS2)
Candida bloodstream infections. Fungal dysbiosis is also linked regions of fungal ribosomal DNA are both common
to the development and treatment response in non-fungal targets in amplicon-based sequencing method and have
infections, for example Clostridioides difficile colitis and HIV. provided similar observations [6–8]: In the healthy human
Further research is needed to define the contribution of gastrointestinal tract, the most common fungal taxa are
intestinal mycobiota to human fungal and non-fungal yeasts such as Candida, Saccharomyces, and Malassezia and
infections. filamentous fungi such as Aspergillus, Cladosporium, and
Penicillium. Collectively, these fungal constituents repre-
Addresses
1 sent core mycobiota species.
Infectious Disease Service, Department of Medicine, Memorial Hospi-
tal, New York, NY, USA
2
Immunology Program, Sloan Kettering Institute, Memorial Sloan Shotgun metagenomics sequencing is another commonly
Kettering Cancer Center, New York, NY, USA used methodology for microbiota analysis and provides
reads of the total microbiota without a potential bias from
Corresponding authors: targeted PCR amplification. Shotgun sequencing of
Rolling, Thierry (rollingt@mskcc.org), Zhai, Bing (zhaib@mskcc.org)
3
These authors contributed equally to this manuscript.
healthy donor samples within the Human Microbiota
Project confirmed the fungal composition obtained by
Current Opinion in Microbiology 2020, 56:1–6 ITS sequencing [7]. Although the presence of many
This review comes from a themed issue on Microbiota
species detected in sequencing data can be verified by
cultivation methods [6], a recent study showed that a
Edited by Iliyan D Iliev
considerable portion of recovered fungal DNA from
healthy human fecal samples were in fact derived from
food or other transient contents [9]. This study corre-
https://doi.org/10.1016/j.mib.2020.05.004 lated a reduction in fecal Candida albicans levels with an
1369-5274/ã 2020 Elsevier Ltd. All rights reserved.
increase in the frequency of oral hygiene. These observa-
tions suggest that core mycobiota species have a higher
volatility than the core bacterial microbiota in the healthy
human gut.

In contrast, in patients with a high risk of fungal infection,


Introduction such as allogeneic hematopoietic cell transplant (allo-
The human intestinal microbiota is a complex reservoir of HCT) recipients, longitudinal sampling of fecal speci-
viruses, bacteria, archaea, fungi, and protozoa. The mens demonstrated that certain Candida species could
impact of the bacterial microbiota on systemic infections, rapidly expand and dominate the intestinal mycobiota,
especially in allogeneic hematopoietic cell transplant which precedes systemic fungal infections [10]. There-
(allo-HCT) patients, has been extensively investigated fore, the intestinal mycobiota is characterized by distinct
[1–5]. In contrast, investigations on the impact of the patterns and constituents under different pathological
fungal compartment of the intestinal microbial flora (i.e. conditions.
mycobiota) have only started recently, in part because
fungi generally represent a minor microbial component in Current limitations of mycobiota analysis
the intestinal niche. In this review, we discuss features of Metagenomics data in analyzing bacterial microbiota
the human intestinal mycobiota and technical challenges provide unbiased read numbers of different taxa. They
in its analysis. We focus on summarizing recent research also allow a functional characterization of the community,

www.sciencedirect.com Current Opinion in Microbiology 2020, 56:1–6

Downloaded for Anonymous User (n/a) at KAWEAH DELTA MEDICAL CENTER from ClinicalKey.com by Elsevier on July 01, 2020.
For personal use only. No other uses without permission. Copyright ©2020. Elsevier Inc. All rights reserved.
2 Microbiota

for example by determining the frequency of drug resis- 16S amplicons in bacteria, the ITS1 amplicons from
tance genes or of genes implicated in metabolic pathways. different fungal species ranges from less than 200 bp to
However, this method is impractical for detailed charac- over 500 bp in length [14]. Since the widely used Illumina
terization of the mycobiota. Multiple studies have shown Miseq platform preferentially sequences smaller DNA
that less than 0.1% of shotgun reads from healthy human fragments [15], fungal species with long ITS regions (e.g.
fecal samples aligned to fungal genomes [7,11], ranging Saccharomyces cerevisiae [16]) can be underrepresented in
from a few hundred to several thousand reads (Figure 1). the final data output. The effect of this underrepresenta-
Even in a fecal sample with a drastic expansion of Candida tion will be lower in samples characterized by exponential
parapsilosis, in which nearly half of the shotgun reads overgrowth of specific fungal species than in samples that
mapped to one dominant fungal species, the fungal contain a diverse mycobiota.
sequences (8.4  104 reads, 8.4 Mb sequence size) did
not amount to a full coverage of the C. parapsilosis genome Although new sequencing strategies, for example full-
(13 Mb). In general, shotgun sequencing fails to provide length rDNA sequencing [17] may bypass the size limita-
sufficient fungal reads for robust compositional or func- tions of the Illumina platform, at the present time ampli-
tional mycobiota analysis. An alternative approach for con-based sequencing is still the best approach to char-
functional analysis would be to isolate the fungus from acterize the intestinal mycobiota. Due to the limitations
fecal samples and to perform whole genome sequencing of the ITS sequencing pipeline and the inherently higher
on the isolates. This approach would bypass the issue of variability of the intestinal mycobiota, a single sampling
sequencing depth with fungal genomes but is only appli- timepoint may not reflect the dynamic nature of intestinal
cable to viable fungal cultures from fecal samples. mycobiota and yield spurious findings in clinical studies.
Instead, researchers should focus on longitudinal collec-
Given the limitations of shotgun metagenomics, current tion of patient samples to provide a dynamic view of
characterization of the mycobiota relies on ITS amplicon- intestinal mycobiota and to reduce sampling noise in the
based methods. Although the strategy of fungal ITS analysis.
sequencing is analogous to the bacterial 16S sequencing,
fungal sequence analyses encounter technical issues Simultaneous analysis of intestinal fungal and
unprecedented in bacterial sequencing analyses. While bacterial flora
the tandem repeats of rDNA operons vary between 1–15 As discussed above, the mycobiota represents a minority
copies in different bacterial species [12], this variation of the total intestinal flora. In most instances, bacteria,
ranges between 14–1442 copies in fungal species [13]. An especially anaerobes, are the major constituents in the
ideal mycobiota pipeline would normalize the relative intestinal niche. Researchers have associated the pres-
abundance of each species according to the rDNA copy ence of anaerobes with intestinal colonization resistance
numbers. However, this normalization is problematic to fungi, particularly Candida species. Consistent with this
given the lack of a reference copy number for all fungal idea, prior exposure to broad-spectrum anti-bacterial anti-
species. Meanwhile, unlike the largely uniform length of biotics increased the risk of developing candidemia [18].
In a murine model, administration of antibiotics was
Figure 1 required for intestinal C. albicans colonization and dis-
semination [19]. A follow-up study showed that the
anaerobic bacteria, Blautia producta (Family: Lachnospir-
108
non-Fungi aceae) and Bacteroides thetaiotamicron (Phylum: Bacteroi-
107 detes), conferred colonization resistance against C. albicans
Fungi
106 [20]. Clinical datasets agreed with these laboratory stud-
# of Reads

105 ies and demonstrated that human intestinal samples with


Candida species expansion exhibited a significant reduc-
104 tion in these bacterial taxa [10,21,22]. These findings
103 point to cross-kingdom interactions between bacteria and
102 fungi in the intestinal microbiota. Therefore, it is crucial
101
to analyze fungal and bacterial dynamics in parallel to
assess complementary and antagonistic functions of
100 members of both kingdoms in the same ecologic niche.
A B
Current Opinion in Microbiology
The mycobiota and fungal bloodstream
infections
The fungal reads and non-fungal reads in metagenomic sequencing Candida species cause over 20 000 bloodstream infection
data of human fecal samples (A) without Candida domination, and (B)
with Candida domination. Sample B was collected from a patient with
cases per year in the United States alone [23]. Even with
active candidemia. The fungal reads account for <0.01% of total contemporary antifungal drugs, fungus-attributable mor-
reads in sample A, and close to 50% of total reads in sample B. tality is higher than 25% [23,24]. Patients in intensive care

Current Opinion in Microbiology 2020, 56:1–6 www.sciencedirect.com

Downloaded for Anonymous User (n/a) at KAWEAH DELTA MEDICAL CENTER from ClinicalKey.com by Elsevier on July 01, 2020.
For personal use only. No other uses without permission. Copyright ©2020. Elsevier Inc. All rights reserved.
The intestinal mycobiota and Systemic Infections Rolling, Hohl and Zhai 3

units and immunosuppressed patients, especially those Figure 2


with prolonged neutropenia, are at an increased risk of
invasive candidiasis [25]. Early identification of high-risk Anti-anaerobic antibiotcs
patients and preemptive treatment may reduce Candida-
related morbidity and mortality [25]. Fungi

Relative abundance
The observation that ingestion of a high dose of live C.
albicans leads to candidemia in a healthy volunteer raised Fungal BSI
the hypothesis of an intestinal source for Candida blood-
stream infections [26]. Further studies showed that Can-
dida bloodstream infections are associated with a high
absolute Candida intestinal burden, and that Candida can Anaerobes
be detected in stool samples before the development of a Time
bloodstream infection in neutropenic patients and in Current Opinion in Microbiology
neonates (systematically reviewed in Ref. [27]).
The dynamics of intestinal fungal and bacterial flora in the context of
The intestinal Candida reservoir was confirmed to be an fungal bloodstream infection. This abstractive figure was plotted by re-
important source for Candida bloodstream infections by analyzing data from a previously published study [10].
ITS1 sequencing of longitudinally collected fecal sam-
ples in a case-control studies of allo-HCT patients [10].
All but one of patient in the Candida bloodstream infec-
tion group experienced a marked intestinal expansion, This hypothesis requires further exploration in relevant
both in terms of relative and absolute abundance, of clinical studies.
corresponding Candida species (C. parapsilosis complex
species and C. albicans) before bloodstream infection. The mycobiota and C. difficile infection
Similar levels of fungal dysbiosis were not observed in C. difficile is the main cause of nosocomially acquired
control patients during the study period. Whole genome diarrhea (C. difficile infection, CDI) [30,31]. Broad-spec-
sequencing of recovered Candida isolates confirmed the trum antibiotic use and the ensuing disturbances in the
similarity of strains found in paired stool and blood bacterial microbiota predispose patients to CDI [32,33].
samples from individual patients. Therefore, the intesti- Three studies have shown differential enrichment of
nal Candida expansion could serve as a biomarker to help multiple fungal taxa in patients with CDI by association
identify patients at high risk for systemic candidiasis. analyses [34–36]. A limitation of these studies is their low
sample size (10–18 patients with CDI) and the possibility
This unusual expansion of specific Candida species dur- of an inflated type I error due to multiple testing. Addi-
ing allo-HCT was influenced by multiple factors. First, tionally, the clinical relevance of the reported taxa enrich-
the expansion of Candida was associated with the loss of ment (such as Helotiales and Byssochlamys) is unclear, as
the intestinal bacterial density and diversity, especially of they are not generally considered as part of the core
the anaerobes. The dysbiosis of the anaerobic community mycobiota [37].
was linked to the prolonged administration of broad-
spectrum antibiotics (Figure 2). Second, C. parapsilosis A randomized controlled trial of fecal microbiota transfer
complex species were frequently observed as the driver of (FMT) for the treatment of CDI showed that CDI
intestinal domination and causative agent of bloodstream patients had a higher baseline (before FMT) intestinal
infections in this cohort. This predominance may be fungal load with lower fungal alpha-diversity than healthy
associated with the use of antifungal prophylaxis with controls, mainly due to an enrichment in C. albicans [38].
micafungin during the first week of allo-HCT, since C. FMT non-responders had a lower fungal diversity with a
parapsilosis complex species are more resistant to mica- higher abundance of C. albicans compared to responders
fungin than other Candida species [28]. (post FMT). This finding was possibly linked to a reduc-
tion in bacterial taxa that conferred Candida colonization
Beyond external factors such as administration of anti- resistance (specifically Lachnospiraceae and Bacteroidetes
biotics, a recent study proposed a pathogenic synergy [20]) in the non-responders. Similarly, in a study compar-
between bacterial and fungal pathogens at the onset of ing fidaxomicin to vancomycin for CDI, Candida intesti-
systemic candidiasis [29]. In a murine model, C. albicans nal colonization was more frequently observed in the
gavage and colonization induced a reduction in oral and vancomycin arm [39], since fidaxomicin and not vanco-
intestinal bacterial diversity, specifically with an over- mycin was known to spare Bacteroides [40]. Collectively,
growth of Enterococcus spp. in both sites. This observation these findings were consistent with idea that C. albicans is
suggested that a high burden of Enterococci diminished not directly associated with CDI but rather represents a
oral mucosal integrity and facilitated C. albicans invasion. marker for bacterial dysbiosis observed in CDI.

www.sciencedirect.com Current Opinion in Microbiology 2020, 56:1–6

Downloaded for Anonymous User (n/a) at KAWEAH DELTA MEDICAL CENTER from ClinicalKey.com by Elsevier on July 01, 2020.
For personal use only. No other uses without permission. Copyright ©2020. Elsevier Inc. All rights reserved.
4 Microbiota

On the other hand, administration of C. albicans prior or  of special interest


 of outstanding interest
concomitant with FMT reduced FMT efficacy in a
murine model of CDI [38]. This result suggests that 1. Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A,
intestinal colonized C. albicans may directly influence the Lee YJ, Dubin KA, Socci ND, Viale A et al.: Intestinal domination
and the risk of bacteremia in patients undergoing allogeneic
efficacy of FMT. In another study, pre-colonization with hematopoietic stem cell transplantation. Clin Infect Dis 2012,
C. albicans prolonged mouse survival after induction of 55:905-914.
CDI, without affecting C. difficile growth or toxin titers. 2. Lee YJ, Arguello ES, Jenq RR, Littmann E, Kim GJ, Miller LC,
The improved survival was linked to an enhanced pro- Ling L, Figueroa C, Robilotti E, Perales MA et al.: Protective
factors in the intestinal microbiome against Clostridium
tective IL-17 response in Candida-colonized mice [41]. difficile infection in recipients of allogeneic hematopoietic
These mouse data suggest differential effects of C. albi- stem cell transplantation. J Infect Dis 2017, 215:1117-1123.
cans on CDI outcomes, based on the timing of Candida 3. Tamburini FB, Andermann TM, Tkachenko E, Senchyna F,
administration and study setting and will require valida- Banaei N, Bhatt AS: Precision identification of diverse
bloodstream pathogens in the gut microbiome. Nat Med 2018,
tion in human cohorts. 24:1809-1814.
4. Kim SG, Becattini S, Moody TU, Shliaha PV, Littmann ER, Seok R,
The mycobiota and HIV infection Gjonbalaj M, Eaton V, Fontana E, Amoretti L et al.: Microbiota-
Advanced HIV infection with low CD4 T-cell counts is a derived lantibiotic restores resistance against vancomycin-
resistant Enterococcus. Nature 2019, 572:665-669.
known risk factor for opportunistic fungal infections.
Several studies suggest that patients infected with HIV 5. Stoma I, Littmann ER, Peled JU, Giralt S, van den Brink MRM,
Pamer EG, Taur Y: Compositional flux within the intestinal
have increased serum levels of beta-D-glucan (BDG, a microbiota and risk for bloodstream infection with gram-
fungal cell wall component) compared to non-infected negative bacteria. Clin Infect Dis 2020. Online ahead of print.
controls [42,43]. Elevated BDG levels are associated 6. Strati F, Di Paola M, Stefanini I, Albanese D, Rizzetto L, Lionetti P,
with immune activation and high circulating inflamma- Calabro A, Jousson O, Donati C, Cavalieri D et al.: Age and gender
affect the composition of fungal population of the human
tory markers in these patients [44,45]. This observation gastrointestinal tract. Front Microbiol 2016, 7:1227.
led to the hypothesis that fungal translocation from the
7. Nash AK, Auchtung TA, Wong MC, Smith DP, Gesell JR, Ross MC,
gut or other mucosal surfaces could lead to immune Stewart CJ, Metcalf GA, Muzny DM, Gibbs RA et al.: The gut
activation and disease progression in HIV patients, simi- mycobiome of the human microbiome project healthy cohort.
larly to what has been postulated for bacteria [46,47]. This Microbiome 2017, 5:153.
hypothesis will need further validation due to a lack of 8. Ward TL, Dominguez-Bello MG, Heisel T, Al-Ghalith G, Knights D,
data on the intestinal mycobiota in HIV patients [48]. Gale CA: Development of the human mycobiome over the first
month of life and across body sites. mSystems 2018, 3.
9. Auchtung TA, Fofanova TY, Stewart CJ, Nash AK, Wong MC,
Future outlook  Gesell JR, Auchtung JM, Ajami NJ, Petrosino JF: Investigating
For many gastrointestinal infections, such as spontaneous colonization of the healthy adult gastrointestinal tract by
peritonitis [49], cholecystitis [50], and superinfected pan- fungi. mSphere 2018, 3.
This article explored the composition of the intestinal mycobiota in
creatic pseudocysts [51], fungi are well-known causative healthy volunteers, and examined the impact of food intake and other
pathogens. The gastrointestinal mycobiota appears to be transient factors on mycobiota sequencing results.
a likely source for many of these infections. Longitudinal 10. Zhai B, Ola M, Rolling T, Tosini NL, Joshowitz S, Littmann ER,
sampling of intestinal or fecal specimens from these  Amoretti LA, Fontana E, Wright RJ, Miranda E et al.: High-
resolution mycobiota analysis reveals dynamic intestinal
patients may facilitate monitoring mycobiota dynamics translocation preceding invasive candidiasis. Nat Med 2020,
and may help provide more definitive answers about a 26:59-64.
This article showed the dynamics of the intestinal mycobiota and micro-
causal link. In the future, it will be imperative to analyze biota in allo-HCT patients and demonstrated that the intestinal mycobiota
the intestinal mycobiota in parallel with bacterial flora to is the source of fungal bloodstream infections.
reveal comprehensive cross-kingdom microbial interac- 11. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-
tions in this complex niche and to define their impact on Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN,
Anokhin AP et al.: Human gut microbiome viewed across age
human disease development. and geography. Nature 2012, 486:222-227.
12. Vetrovsky T, Baldrian P: The variability of the 16S rRNA gene in
Conflict of interest statement bacterial genomes and its consequences for bacterial
Nothing declared. community analyses. PLoS One 2013, 8:e57923.
13. Lofgren LA, Uehling JK, Branco S, Bruns TD, Martin F,
Acknowledgements Kennedy PG: Genome-based estimates of fungal rDNA copy
This work was supported by a Geoffrey Beene Foundation Award (to T.M. number variation across phylogenetic scales and ecological
lifestyles. Mol Ecol 2019, 28:721-730.
H.), a Burroughs Wellcome Fund Investigator in the Pathogenesis of
Infectious Diseases Award (to T.M.H.), National Institutes of Health grants 14. Tang J, Iliev ID, Brown J, Underhill DM, Funari VA: Mycobiome:
(nos. P30 CA008748 to the MSKCC, R01 AI093808 and R01 AI 139632 to approaches to analysis of intestinal fungi. J Immunol Methods
T.M.H.), and a Deutsche Forschungsgemeinschaft (German Research 2015, 421:112-121.
Foundation) fellowship grant (no. RO5328/2-1 to T.R.).
15. Gohl DM, Magli A, Garbe J, Becker A, Johnson DM, Anderson S,
Auch B, Billstein B, Froehling E, McDevitt SL et al.: Measuring
References and recommended reading sequencer size bias using REcount: a novel method for highly
Papers of particular interest, published within the period of review, accurate Illumina sequencing-based quantification. Genome
have been highlighted as: Biol 2019, 20:85.

Current Opinion in Microbiology 2020, 56:1–6 www.sciencedirect.com

Downloaded for Anonymous User (n/a) at KAWEAH DELTA MEDICAL CENTER from ClinicalKey.com by Elsevier on July 01, 2020.
For personal use only. No other uses without permission. Copyright ©2020. Elsevier Inc. All rights reserved.
The intestinal mycobiota and Systemic Infections Rolling, Hohl and Zhai 5

16. Motooka D, Fujimoto K, Tanaka R, Yaguchi T, Gotoh K, Maeda Y, microbiome in recurrent Clostridium difficile-associated
Furuta Y, Kurakawa T, Goto N, Yasunaga T et al.: Fungal ITS1 diarrhea. J Infect Dis 2008, 197:435-438.
deep-sequencing strategies to reconstruct the composition of
a 26-species community and evaluation of the gut mycobiota 34. Lamendella R, Wright JR, Hackman J, McLimans C, Toole DR,
of healthy Japanese individuals. Front Microbiol 2017, 8:238. Bernard Rubio W, Drucker R, Wong HT, Sabey K, Hegarty JP et al.:
Antibiotic treatments for Clostridium difficile infection are
17. Callahan BJ, Wong J, Heiner C, Oh S, Theriot CM, Gulati AS, associated with distinct bacterial and fungal community
McGill SK, Dougherty MK: High-throughput amplicon structures. mSphere 2018, 3.
sequencing of the full-length 16S rRNA gene with single-
nucleotide resolution. Nucleic Acids Res 2019, 47:e103. 35. Stewart DB Sr, Wright JR, Fowler M, McLimans CJ, Tokarev V,
Amaniera I, Baker O, Wong HT, Brabec J, Drucker R et al.:
18. Yapar N: Epidemiology and risk factors for invasive Integrated meta-omics reveals a fungus-associated
candidiasis. Ther Clin Risk Manag 2014, 10:95-105. bacteriome and distinct functional pathways in Clostridioides
difficile infection. mSphere 2019, 4.
19. Koh AY, Kohler JR, Coggshall KT, Van Rooijen N, Pier GB:
Mucosal damage and neutropenia are required for Candida 36. Sangster W, Hegarty JP, Schieffer KM, Wright JR, Hackman J,
albicans dissemination. PLoS Pathog 2008, 4:e35. Toole DR, Lamendella R, Stewart DB Sr: Bacterial and fungal
microbiota changes distinguish C. difficile infection from
20. Fan D, Coughlin LA, Neubauer MM, Kim J, Kim MS, Zhan X, other forms of diarrhea: results of a prospective inpatient
Simms-Waldrip TR, Xie Y, Hooper LV, Koh AY: Activation of study. Front Microbiol 2016, 7:789.
HIF-1alpha and LL-37 by commensal bacteria inhibits Candida
albicans colonization. Nat Med 2015, 21:808-814. 37. Fiers WD, Gao IH, Iliev ID: Gut mycobiota under scrutiny: fungal
symbionts or environmental transients? Curr Opin Microbiol
21. Hoarau G, Mukherjee PK, Gower-Rousseau C, Hager C, 2019, 50:79-86.
Chandra J, Retuerto MA, Neut C, Vermeire S, Clemente J,
Colombel JF et al.: Bacteriome and mycobiome interactions 38. Zuo T, Wong SH, Cheung CP, Lam K, Lui R, Cheung K, Zhang F,
underscore microbial dysbiosis in familial Crohn’s disease.  Tang W, Ching JYL, Wu JCY et al.: Gut fungal dysbiosis
mBio 2016, 7. correlates with reduced efficacy of fecal microbiota
transplantation in Clostridium difficile infection. Nat Commun
22. Leonardi I, Paramsothy S, Doron I, Semon A, Kaakoush NO, 2018, 9:3663.
Clemente JC, Faith JJ, Borody TJ, Mitchell HM, Colombel JF et al.: This study showed an increased relative abundance of Candida spp. In
Fungal trans-kingdom dynamics linked to responsiveness to the ‘nonresponders’ of CDI patients that received a fecal microbiota
Fecal Microbiota Transplantation (FMT) therapy in ulcerative transplant.
colitis. Cell Host Microbe 2020, 27 823-829.e3.
39. Nerandzic MM, Mullane K, Miller MA, Babakhani F, Donskey CJ:
23. Tsay SV, Mu Y, Williams S, Epson E, Nadle J, Bamberg WM, Reduced acquisition and overgrowth of vancomycin-resistant
Barter DM, Johnston HL, Farley MM, Harb S et al.: Burden of enterococci and Candida species in patients treated with
candidemia in the United States, 2017. Clin Infect Dis 2020. fidaxomicin versus vancomycin for Clostridium difficile
Online ahead of print. infection. Clin Infect Dis 2012, 55(Suppl. 2):S121-S126.
24. Mejia-Chew C, O’Halloran JA, Olsen MA, Stwalley D, Kronen R, 40. Louie TJ, Emery J, Krulicki W, Byrne B, Mah M: OPT-80
Lin C, Salazar AS, Larson L, Hsueh K, Powderly WG et al.: Effect of eliminates Clostridium difficile and is sparing of bacteroides
infectious disease consultation on mortality and treatment of species during treatment of C. difficile infection. Antimicrob
patients with candida bloodstream infections: a retrospective, Agents Chemother 2009, 53:261-263.
cohort study. Lancet Infect Dis 2019, 19:1336-1344.
41. Markey L, Shaban L, Green ER, Lemon KP, Mecsas J,
25. Kullberg BJ, Arendrup MC: Invasive candidiasis. N Engl J Med Kumamoto CA: Pre-colonization with the commensal fungus
2015, 373:1445-1456. Candida albicans reduces murine susceptibility to Clostridium
difficile infection. Gut Microbes 2018, 9:497-509.
26. Krause W, Matheis H, Wulf K: Fungaemia and funguria after oral
administration of Candida albicans. Lancet 1969, 1:598-599. 42. Dirajlal-Fargo S, El-Kamari V, Weiner L, Shan L, Sattar A,
 Kulkarni M, Funderburg N, Nazzinda R, Karungi C, Kityo C et al.:
27. Nucci M, Anaissie E: Revisiting the source of candidemia: skin Altered intestinal permeability and fungal translocation in
or gut? Clin Infect Dis 2001, 33:1959-1967. Ugandan children with HIV. Clin Infect Dis 2019, 70:2413-2422.
This study showed evidence of fungal translocation from gut mycobiota in
28. Garcia-Effron G, Katiyar SK, Park S, Edlind TD, Perlin DS: HIV patients.
A naturally occurring proline-to-alanine amino acid change
in Fks1p in Candida parapsilosis, Candida orthopsilosis, 43. Weiner LD, Retuerto M, Hager CL, El Kamari V, Shan L, Sattar A,
and Candida metapsilosis accounts for reduced Kulkarni M, Funderburg N, Ghannoum MA, Dirajlal-Fargo S et al.:
echinocandin susceptibility. Antimicrob Agents Chemother Fungal translocation is associated with immune activation
2008, 52:2305-2312. and systemic inflammation in treated HIV. AIDS Res Hum
Retroviruses 2019, 35:461-472.
29. Bertolini M, Ranjan A, Thompson A, Diaz PI, Sobue T, Maas K,
 Dongari-Bagtzoglou A: Candida albicans induces mucosal 44. Hoenigl M, Moser CB, Funderburg N, Bosch R, Kantor A, Zhang Y,
bacterial dysbiosis that promotes invasive infection. PLoS Eugen-Olsen J, Finkelman M, Reiser J, Landay A et al.: Soluble
Pathog 2019, 15:e1007717. urokinase plasminogen activator receptor is predictive of non-
This article proposed a synergistic model of pathogenesis between Can- AIDS events during antiretroviral therapy-mediated viral
dida albicans and pathogenic bacteria in mice. suppression. Clin Infect Dis 2019, 69:676-686.
30. Magill SS, Edwards JR, Bamberg W, Beldavs ZG, Dumyati G, 45. Hoenigl M, de Oliveira MF, Perez-Santiago J, Zhang Y, Woods SP,
Kainer MA, Lynfield R, Maloney M, McAllister-Hollod L, Nadle J Finkelman M, Gianella S: Correlation of (1!3)-beta-D-glucan
et al.: Multistate point-prevalence survey of health care- with other inflammation markers in chronically HIV infected
associated infections. N Engl J Med 2014, 370:1198-1208. persons on suppressive antiretroviral therapy. GMS Infect Dis
2015, 3.
31. Manthey CF, Dranova D, Christner M, Berneking L, Kluge S,
Lohse AW, Fuhrmann V: Stool cultures at the ICU: get rid of it! 46. Klatt NR, Funderburg NT, Brenchley JM: Microbial translocation,
Ann Intensive Care 2018, 8:10. immune activation, and HIV disease. Trends Microbiol 2013,
21:6-13.
32. Brown KA, Khanafer N, Daneman N, Fisman DN: Meta-analysis
of antibiotics and the risk of community-associated 47. Hoenigl M: Fungal translocation: a driving force behind the
Clostridium difficile infection. Antimicrob Agents Chemother occurrence of non-AIDS events? Clin Infect Dis 2020,
2013, 57:2326-2332. 70:242-244.
33. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT, 48. Hager CL, Ghannoum MA: The mycobiome in HIV. Curr Opin HIV
Schmidt TM, Young VB: Decreased diversity of the fecal AIDS 2018, 13:69-72.

www.sciencedirect.com Current Opinion in Microbiology 2020, 56:1–6

Downloaded for Anonymous User (n/a) at KAWEAH DELTA MEDICAL CENTER from ClinicalKey.com by Elsevier on July 01, 2020.
For personal use only. No other uses without permission. Copyright ©2020. Elsevier Inc. All rights reserved.
6 Microbiota

49. Tariq T, Irfan FB, Farishta M, Dykstra B, Sieloff EM, Desai AP: prospective study comparing next generation sequencing to
Spontaneous fungal peritonitis: micro-organisms, culture. J Infect 2020, 80:16-23.
management and mortality in liver cirrhosis-a systematic
review. World J Hepatol 2019, 11:596-606. 51. Reuken PA, Albig H, Rodel J, Hocke M, Will U, Stallmach A,
Bruns T: Fungal infections in patients with infected pancreatic
50. Dyrhovden R, Ovrebo KK, Nordahl MV, Nygaard RM, Ulvestad E, necrosis and pseudocysts: risk factors and outcome. Pancreas
Kommedal O: Bacteria and fungi in acute cholecystitis. A 2018, 47:92-98.

Current Opinion in Microbiology 2020, 56:1–6 www.sciencedirect.com

Downloaded for Anonymous User (n/a) at KAWEAH DELTA MEDICAL CENTER from ClinicalKey.com by Elsevier on July 01, 2020.
View publication stats For personal use only. No other uses without permission. Copyright ©2020. Elsevier Inc. All rights reserved.

You might also like